1
|
Kores K, Lešnik S, Bren U. Computational Analysis of S1PR1 SNPs Reveals Drug Binding Modes Relevant to Multiple Sclerosis Treatment. Pharmaceutics 2024; 16:1413. [PMID: 39598537 PMCID: PMC11597183 DOI: 10.3390/pharmaceutics16111413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS) characterized by myelin and axonal damage with a globally rising incidence. While there is no known cure for MS, various disease-modifying treatments (DMTs) exist, including those targeting Sphingosine-1-Phosphate Receptors (S1PRs), which play important roles in immune response, CNS function, and cardiovascular regulation. This study focuses on understanding how nonsynonymous single nucleotide polymorphisms (rs1299231517, rs1323297044, rs1223284736, rs1202284551, rs1209378712, rs201200746, and rs1461490142) in the S1PR1's active site affect the binding of endogenous ligands, as well as different drugs used in MS management. Methods: Extensive molecular dynamics simulations and linear interaction energy (LIE) calculations were employed to predict binding affinities and potentially guide future personalized medicinal therapies. The empirical parameters of the LIE method were optimized using the binding free energies calculated from experimentally determined IC50 values. These optimized parameters were then applied to calculate the binding free energies of S1P to mutated S1PR1, which correlated well with experimental values, confirming their validity for assessing the impact of SNPs on S1PR1 binding affinities. Results: The binding free energies varied from the least favorable -8.2 kcal/mol for the wild type with ozanimod to the most favorable -16.7 kcal/mol for the combination of siponimod with the receptor carrying the F2055.42L mutation. Conclusions: We successfully demonstrated the differences in the binding modes, interactions, and affinities of investigated MS drugs in connection with SNPs in the S1PR1 binding site, resulting in several viable options for personalized therapies depending on the present mutations.
Collapse
Grants
- P2-0046, P1-0403, J1-2471, L2-3175, P2-0438, J1-4398, L2-4430, J3-4498, J7-4638, J1-421 4414, J3-4497, J4-4633, J1-50034, J7-50034, I0-E015 Slovenian Research and Innovation Agency (ARIS)
Collapse
Affiliation(s)
- Katarina Kores
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (S.L.)
| | - Samo Lešnik
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (S.L.)
- Institute of Environmental Protection and Sensors, IOS, Beloruska 7, SI-2000 Maribor, Slovenia
| | - Urban Bren
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia; (K.K.); (S.L.)
- Institute of Environmental Protection and Sensors, IOS, Beloruska 7, SI-2000 Maribor, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000 Koper, Slovenia
| |
Collapse
|
2
|
Maruyama Y, Ohsawa Y, Suzuki T, Yamauchi Y, Ohno K, Inoue H, Yamamoto A, Hayashi M, Okuhara Y, Muramatsu W, Namiki K, Hagiwara N, Miyauchi M, Miyao T, Ishikawa T, Horie K, Hayama M, Akiyama N, Hirokawa T, Akiyama T. Pseudoirreversible inhibition elicits persistent efficacy of a sphingosine 1-phosphate receptor 1 antagonist. Nat Commun 2024; 15:5743. [PMID: 39030171 PMCID: PMC11271513 DOI: 10.1038/s41467-024-49893-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/19/2024] [Indexed: 07/21/2024] Open
Abstract
Sphingosine 1-phosphate receptor 1 (S1PR1), a G protein-coupled receptor, is required for lymphocyte trafficking, and is a promising therapeutic target in inflammatory diseases. Here, we synthesize a competitive S1PR1 antagonist, KSI-6666, that effectively suppresses pathogenic inflammation. Metadynamics simulations suggest that the interaction of KSI-6666 with a methionine residue Met124 in the ligand-binding pocket of S1PR1 may inhibit the dissociation of KSI-6666 from S1PR1. Consistently, in vitro functional and mutational analyses reveal that KSI-6666 causes pseudoirreversible inhibition of S1PR1, dependent on the Met124 of the protein and substituents on the distal benzene ring of KSI-6666. Moreover, in vivo study suggests that this pseudoirreversible inhibition is responsible for the persistent activity of KSI-6666.
Collapse
Affiliation(s)
- Yuya Maruyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Yusuke Ohsawa
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Takayuki Suzuki
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Yuko Yamauchi
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Kohsuke Ohno
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Hitoshi Inoue
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Akitoshi Yamamoto
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Morimichi Hayashi
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Yuji Okuhara
- Central Research Laboratory, Kissei Pharmaceutical Co., Ltd., 4365-1 Hotaka-Kashiwabara, Azumino, Nagano, 399-8304, Japan
| | - Wataru Muramatsu
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Kano Namiki
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Naho Hagiwara
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Maki Miyauchi
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Takahisa Miyao
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Tatsuya Ishikawa
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Kenta Horie
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Mio Hayama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Nobuko Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Takatsugu Hirokawa
- Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Transborder Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Taishin Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan.
| |
Collapse
|
3
|
Kurano M, Uranbileg B, Yatomi Y. Apolipoprotein M bound sphingosine 1-phosphate suppresses NETosis through activating S1P1 and S1P4. Biomed Pharmacother 2023; 166:115400. [PMID: 37657263 DOI: 10.1016/j.biopha.2023.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
The pleiotropic effects of high-density lipoprotein (HDL), including its protective properties against sepsis, are attributed to the sphingosine 1-phosphate and apolipoprotein M (ApoM) that are carried on the lipoproteins. In this study, we attempted to elucidate the possible mechanisms underlying the sepsis coagulopathic state by considering the modulation of NETosis. Our results revealed that in a lipopolysaccharide-induced sepsis mouse model, the levels of NETosis markers, such as plasma DNA and histone, were elevated in ApoM-knockout (KO) mice and attenuated in ApoM-overexpressing mice. In ApoM-KO mice, the survival rate decreased and the occurrence rates of coagulopathy and organ injury increased following the administration of histone. Treatment with a conditioned medium of ApoM-overexpressing cells attenuated the observed NETosis in HL-60S cells that differentiated into neutrophils and were inhibited through the suppression of S1P1 or S1P4. The attenuation of PKCδ and PKCα/β by S1P1 and S1P4 activation may also be involved. In ApoM-overexpressing mice, coagulopathy and organ injuries were attenuated following an injection of histone; these effects were partially inhibited by S1P1, 3, S1P4, or S1P1 antagonists. Furthermore, the exogenous administration of ApoM protected ApoM-KO mice that were challenged with histone from developing NETosis. In conclusion, the ApoM/S1P axis protects against NETosis through the attenuation of PKC activation by S1P1 and S1P4. The development of drugs targeting the ApoM/S1P axis may be beneficial for the treatment of pathological conditions involving uncontrolled NETosis, such as sepsis.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan.
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Kurano M, Tsukamoto K, Shimizu T, Hara M, Yatomi Y. Apolipoprotein M/sphingosine 1-phosphate protects against diabetic nephropathy. Transl Res 2023:S1931-5244(23)00024-5. [PMID: 36805561 DOI: 10.1016/j.trsl.2023.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/10/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023]
Abstract
Diabetic nephropathy remains a common cause of end-stage renal failure and its associated mortality around the world. Sphingosine 1-phosphate (S1P) is a multifunctional lipid mediator and binds to HDL via apolipoprotein M (ApoM). Since HDL has been reported to be epidemiologically associated with kidney disease, we attempted to investigate the involvement of the ApoM/S1P axis in the pathogenesis/progression of diabetic nephropathy. In type 2 diabetic patients, the serum ApoM levels were inversely correlated with the clinical stage of diabetic nephropathy. The decline in the eGFR over a 5-year observation period proceeded more rapidly in subjects with lower serum ApoM levels. In a mouse model of streptozotocin-induced diabetes, deletion of ApoM deteriorated the phenotypes of diabetic nephropathy: the urinary albumin and plasma creatinine levels increased, the kidneys enlarged, and renal fibrosis and thickening of the basement membrane progressed. On the other hand, overexpression of ApoM ameliorated these phenotypes. These protective effects of ApoM were partially inhibited by treatment with VPC23019, an antagonist of S1P1 and S1P3, but not by treatment with JTE013, an antagonist of S1P2. ApoM/S1P axis attenuated activation of the Smad3 pathway, while augmented eNOS phosphorylation through the S1P1 pathway. Moreover, ApoM/S1P increased the SIRT1 protein levels and enhanced mitochondrial functions by increasing the S1P content of the cell membrane, which might cause selective activation of S1P1. ApoM might be a useful biomarker for predicting the progression of diabetic nephropathy, and the ApoM/S1P-S1P1 axis might serve as a novel therapeutic target for preventing the development/progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine and 5Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Tomo Shimizu
- Tsukuba Research Institute, Research & Development Division, Sekisui Medical Co., Ltd., Ibaraki, Japan
| | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine, Kanagawa, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine and 5Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Chen H, Wang J, Zhang C, Ding P, Tian S, Chen J, Ji G, Wu T. Sphingosine 1-phosphate receptor, a new therapeutic direction in different diseases. Biomed Pharmacother 2022; 153:113341. [PMID: 35785704 DOI: 10.1016/j.biopha.2022.113341] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/01/2022] Open
Abstract
Sphingosine 1-phosphate receptor (S1PR), as a kind of G protein-coupled receptor, has five subtypes, including S1PR1, S1PR2, S1PR3, S1PR4, and S1PR5. Sphingosine 1-phosphate receptor (S1P) and S1PR regulate the trafficking of neutrophils and some cells, which has great effects on immune systems, lung tissue, and liver tissue. Presently, many related reports have proved that S1PR has a strong effect on the migration of lymphocytes, tumor cells, neutrophils, and many other cells via the regulation of signals, pathways, and enzymes. In this way, S1PR can regulate the relative response of the organism. Thus, S1PR has become a possible target for the treatment of autoimmune diseases, pulmonary disease, liver disease, and cancer. In this review, we mainly focus on the research of the S1PR for the new therapeutic directions of different diseases and is expected to assist support in the clinic and drug use.
Collapse
Affiliation(s)
- Hongyu Chen
- Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Caiyun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peilun Ding
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuxia Tian
- Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Junming Chen
- Minhang Hospital, Fudan University, Shanghai 201199, China.
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Lyapina E, Marin E, Gusach A, Orekhov P, Gerasimov A, Luginina A, Vakhrameev D, Ergasheva M, Kovaleva M, Khusainov G, Khorn P, Shevtsov M, Kovalev K, Bukhdruker S, Okhrimenko I, Popov P, Hu H, Weierstall U, Liu W, Cho Y, Gushchin I, Rogachev A, Bourenkov G, Park S, Park G, Hyun HJ, Park J, Gordeliy V, Borshchevskiy V, Mishin A, Cherezov V. Structural basis for receptor selectivity and inverse agonism in S1P 5 receptors. Nat Commun 2022; 13:4736. [PMID: 35961984 PMCID: PMC9374744 DOI: 10.1038/s41467-022-32447-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
The bioactive lysophospholipid sphingosine-1-phosphate (S1P) acts via five different subtypes of S1P receptors (S1PRs) - S1P1-5. S1P5 is predominantly expressed in nervous and immune systems, regulating the egress of natural killer cells from lymph nodes and playing a role in immune and neurodegenerative disorders, as well as carcinogenesis. Several S1PR therapeutic drugs have been developed to treat these diseases; however, they lack receptor subtype selectivity, which leads to side effects. In this article, we describe a 2.2 Å resolution room temperature crystal structure of the human S1P5 receptor in complex with a selective inverse agonist determined by serial femtosecond crystallography (SFX) at the Pohang Accelerator Laboratory X-Ray Free Electron Laser (PAL-XFEL) and analyze its structure-activity relationship data. The structure demonstrates a unique ligand-binding mode, involving an allosteric sub-pocket, which clarifies the receptor subtype selectivity and provides a template for structure-based drug design. Together with previously published S1PR structures in complex with antagonists and agonists, our structure with S1P5-inverse agonist sheds light on the activation mechanism and reveals structural determinants of the inverse agonism in the S1PR family.
Collapse
Affiliation(s)
- Elizaveta Lyapina
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Egor Marin
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Anastasiia Gusach
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Philipp Orekhov
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | | | - Aleksandra Luginina
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Daniil Vakhrameev
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Margarita Ergasheva
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Margarita Kovaleva
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Georgii Khusainov
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- Division of Biology and Chemistry, Paul Scherrer Institute, Forschungsstrasse 111, 5232, Villigen, PSI, Switzerland
| | - Polina Khorn
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Mikhail Shevtsov
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Kirill Kovalev
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- European Molecular Biology Laboratory, Hamburg unit c/o DESY, Hamburg, Germany
| | - Sergey Bukhdruker
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Ivan Okhrimenko
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Petr Popov
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- iMolecule, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, Moscow, 121205, Russia
| | - Hao Hu
- Department of Physics, Arizona State University, Tempe, AZ, 85281, USA
| | - Uwe Weierstall
- Department of Physics, Arizona State University, Tempe, AZ, 85281, USA
| | - Wei Liu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yunje Cho
- Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Ivan Gushchin
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Andrey Rogachev
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
- Joint Institute for Nuclear Research, Dubna, 141980, Russia
| | - Gleb Bourenkov
- European Molecular Biology Laboratory, Hamburg unit c/o DESY, Hamburg, Germany
| | - Sehan Park
- Pohang Accelerator Laboratory, POSTECH, Pohang, 37673, Republic of Korea
| | - Gisu Park
- Pohang Accelerator Laboratory, POSTECH, Pohang, 37673, Republic of Korea
| | - Hyo Jung Hyun
- Pohang Accelerator Laboratory, POSTECH, Pohang, 37673, Republic of Korea
| | - Jaehyun Park
- Pohang Accelerator Laboratory, POSTECH, Pohang, 37673, Republic of Korea
- Department of Chemical Engineering, POSTECH, Pohang, 37673, Republic of Korea
| | - Valentin Gordeliy
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, 38400, France
| | - Valentin Borshchevskiy
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
- Joint Institute for Nuclear Research, Dubna, 141980, Russia.
| | - Alexey Mishin
- Research Сenter for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia.
| | - Vadim Cherezov
- Bridge Institute, Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
7
|
Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition. Cell Res 2021; 31:1263-1274. [PMID: 34526663 PMCID: PMC8441948 DOI: 10.1038/s41422-021-00566-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is an important bioactive lipid molecule in cell membrane metabolism and binds to G protein-coupled S1P receptors (S1PRs) to regulate embryonic development, physiological homeostasis, and pathogenic processes in various organs. S1PRs are lipid-sensing receptors and are therapeutic targets for drug development, including potential treatment of COVID-19. Herein, we present five cryo-electron microscopy structures of S1PRs bound to diverse drug agonists and the heterotrimeric Gi protein. Our structural and functional assays demonstrate the different binding modes of chemically distinct agonists of S1PRs, reveal the mechanical switch that activates these receptors, and provide a framework for understanding ligand selectivity and G protein coupling.
Collapse
|
8
|
Abstract
Sphingosine-1-phosphate (S1P) is a lipidic mediator in mammals that functions either as a second messenger or as a ligand. In the latter case, it is transported by its HDL-associated apoM carrier and circulated in blood where it binds to specific S1P receptors on cell membranes and induces downstream reactions. Although S1P signaling pathways are essential for many biological processes, they are poorly understood at the molecular level. Here, the solved crystal structures of the S1P1 receptor were used to evaluate molecular dynamics (MD) simulations to generate greater detailed molecular insights into the mechanism of S1P signaling. The MD simulations provided observations at the coarse-grained and atomic levels indicating that S1P may access the receptor binding pocket directly from solvents. Lifting of the bulky N-terminal cap region of the receptor precedes initial S1P binding. Glu1213.29 guides S1P penetration, and together with Arg2927.34 is responsible for the stabilization of S1P in the binding pocket, which is consistent with experimental predictions. The complete binding of S1P is followed by receptor activation, wherein Trp2696.48 moves toward the transmembrane helix (TM) 7, resulting in the formation of an enhanced hydrogen bond network in the lower region of TM7. The distance between TM3 and TM6 is subsequently increased, resulting in the opening of the intracellular binding pocket that enables G protein binding. Further analysis of the force distribution network in the receptor yielded a detailed molecular understanding of the signal transmission network that is activated upon agonist binding.
Collapse
|
9
|
Chen Y, Liu T, Xi Q, Jia W, Yin D, Wang X. A Computational Approach to the Study of the Binding Mode of S1P1R Agonists Based on the Active-Like Receptor Model. J Chem Inf Model 2019; 59:1624-1633. [DOI: 10.1021/acs.jcim.8b00764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yonghui Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Tianqi Liu
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Qiumu Xi
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Wenqiang Jia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| |
Collapse
|
10
|
Tigyi GJ, Johnson LR, Lee SC, Norman DD, Szabo E, Balogh A, Thompson K, Boler A, McCool WS. Lysophosphatidic acid type 2 receptor agonists in targeted drug development offer broad therapeutic potential. J Lipid Res 2019; 60:464-474. [PMID: 30692142 PMCID: PMC6399510 DOI: 10.1194/jlr.s091744] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/15/2019] [Indexed: 11/20/2022] Open
Abstract
The growth factor-like lipid mediator, lysophosphatidic acid (LPA), is a potent signaling molecule that influences numerous physiologic and pathologic processes. Manipulation of LPA signaling is of growing pharmacotherapeutic interest, especially because LPA resembles compounds with drug-like features. The action of LPA is mediated through activation of multiple types of molecular targets, including six G protein-coupled receptors that are clear targets for drug development. However, the LPA signaling has been linked to pathological responses that include promotion of fibrosis, atherogenesis, tumorigenesis, and metastasis. Thus, a question arises: Can we harness, in an LPA-like drug, the many beneficial activities of this lipid without eliciting its dreadful actions? We developed octadecyl thiophosphate (OTP; subsequently licensed as Rx100), an LPA mimic with higher stability in vivo than LPA. This article highlights progress made toward developing analogs like OTP and exploring prosurvival and regenerative LPA signaling. We determined that LPA prevents cell death triggered by various cellular stresses, including genotoxic stressors, and rescues cells condemned to apoptosis. LPA2 agonists provide a new treatment option for secretory diarrhea and reduce gastric erosion caused by nonsteroidal anti-inflammatory drugs. The potential uses of LPA2 agonists like OTP and sulfamoyl benzoic acid-based radioprotectins must be further explored for therapeutic uses.
Collapse
Affiliation(s)
- Gabor J Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
- RxBio Inc. Memphis, TN 38163
- Research Division Veterans Affairs Medical Center, Memphis, TN 38104
| | - Leonard R Johnson
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
- RxBio Inc. Memphis, TN 38163
| | - Sue Chin Lee
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
| | - Derek D Norman
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
- Research Division Veterans Affairs Medical Center, Memphis, TN 38104
| | - Erzsebet Szabo
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
| | - Andrea Balogh
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN 38163
| | | | | | | |
Collapse
|
11
|
Blaho VA, Chun J. 'Crystal' Clear? Lysophospholipid Receptor Structure Insights and Controversies. Trends Pharmacol Sci 2018; 39:953-966. [PMID: 30343728 PMCID: PMC6201317 DOI: 10.1016/j.tips.2018.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/11/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022]
Abstract
Lysophospholipids (LPLs), particularly sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA), are bioactive lipid modulators of cellular homeostasis and pathology. The discovery and characterization of five S1P- and six LPA-specific G protein-coupled receptors (GPCRs), S1P1-5 and LPA1-6, have expanded their known involvement in all mammalian physiological systems. Resolution of the S1P1, LPA1, and LPA6 crystal structures has fueled the growing interest in these receptors and their ligands as targets for pharmacological manipulation. In this review, we have attempted to provide an integrated overview of the three crystallized LPL GPCRs with biochemical and physiological structure-function data. Finally, we provide a novel discussion of how chaperones for LPLs may be considered when extrapolating crystallographic and computational data toward understanding actual biological interactions and phenotypes.
Collapse
Affiliation(s)
- Victoria A Blaho
- Sanford Burnham Prebys Medical Discovery Institute, Degenerative Diseases Program, La Jolla, CA 92037, USA.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, Degenerative Diseases Program, La Jolla, CA 92037, USA.
| |
Collapse
|
12
|
Targeting sphingosine-1-phosphate signaling for cancer therapy. SCIENCE CHINA-LIFE SCIENCES 2017. [DOI: 10.1007/s11427-017-9046-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
13
|
Troupiotis-Tsaïlaki A, Zachmann J, González-Gil I, Gonzalez A, Ortega-Gutiérrez S, López-Rodríguez ML, Pardo L, Govaerts C. Ligand chain length drives activation of lipid G protein-coupled receptors. Sci Rep 2017; 7:2020. [PMID: 28515494 PMCID: PMC5435731 DOI: 10.1038/s41598-017-02104-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/07/2017] [Indexed: 01/20/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a lipid mediator that can activate five cell membrane G protein-coupled receptors (GPCRs) which carry a variety of essential functions and are promising drug targets. S1P is composed of a polar zwitterionic head-group and a hydrophobic alkyl chain. This implies an activation mechanism of its cognate receptor that must be significantly different from what is known for prototypical GPCRs (ie receptor to small hydrophilic ligands). Here we aim to identify the structural features responsible for S1P agonism by combining molecular dynamics simulations and functional assays using S1P analogs of different alkyl chain lengths. We propose that high affinity binding involves polar interactions between the lipid head-group and receptor side chains while activation is due to hydrophobic interactions between the lipid tail and residues in a distinct binding site. We observe that ligand efficacy is directly related to alkyl chain length but also varies with receptor subtypes in correlation with the size of this binding pocket. Integrating experimental and computational data, we propose an activation mechanism for the S1P receptors involving agonist-induced conformational events that are conserved throughout class A GPCRs.
Collapse
Affiliation(s)
| | - Julian Zachmann
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Inés González-Gil
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Angel Gonzalez
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Maria L López-Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Cedric Govaerts
- Laboratoire de Structure et Fonction des Membranes Biologiques, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
14
|
Molecular mechanisms of target recognition by lipid GPCRs: relevance for cancer. Oncogene 2015; 35:4021-35. [PMID: 26640151 DOI: 10.1038/onc.2015.467] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022]
|
15
|
Shaikh RS, Keul P, Schäfers M, Levkau B, Haufe G. New fluorinated agonists for targeting the sphingosin-1-phosphate receptor 1 (S1P1). Bioorg Med Chem Lett 2015; 25:5048-51. [DOI: 10.1016/j.bmcl.2015.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/09/2015] [Accepted: 10/10/2015] [Indexed: 11/25/2022]
|
16
|
Archbold JK, Martin JL, Sweet MJ. Towards selective lysophospholipid GPCR modulators. Trends Pharmacol Sci 2014; 35:219-26. [PMID: 24746475 DOI: 10.1016/j.tips.2014.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 01/08/2023]
Abstract
G-protein-coupled receptors (GPCRs) that recognize the lysophospholipids (LPLs) are grouped into two phylogenetically distinct families: the endothelial differentiation gene (Edg) and non-Edg GPCRs. Owing to their more recent identification, and hindered by a lack of selective pharmacological tools, our understanding of the functions and signaling pathways of the non-Edg GPCRs is still in its infancy. Targeting the non-conserved allosteric binding sites of the LPL GPCRs shows particular promise for the development of selective modulators by structure-based drug design. However, only one Edg GPCR (S1PR1) structure has been determined to date, and it has low sequence identity with the non-Edg GPCRs (<20%). Thus, a representative structure of a non-Edg GPCR remains a pressing objective for selective structure-based drug design. Obtaining selective modulators targeting the non-Edg receptors would help to unravel the biology behind these novel GPCRs and potentially will support therapeutic treatment of diseases such as cancer, inflammation, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Julia K Archbold
- Division of Chemistry and Structural Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia.
| | - Jennifer L Martin
- Division of Chemistry and Structural Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
| | - Matthew J Sweet
- Division of Molecular and Cell Biology, The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
| |
Collapse
|
17
|
Tehan BG, Bortolato A, Blaney FE, Weir MP, Mason JS. Unifying family A GPCR theories of activation. Pharmacol Ther 2014; 143:51-60. [PMID: 24561131 DOI: 10.1016/j.pharmthera.2014.02.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/05/2014] [Indexed: 10/25/2022]
Abstract
Several new pairs of active and inactive GPCR structures have recently been solved enabling detailed structural insight into the activation process, not only of rhodopsin but now also of the β2 adrenergic, M2 muscarinic and adenosine A2A receptors. Combined with structural analyses they have enabled us to examine the different recent theories proposed for GPCR activation and show that they are all indeed parts of the same process, and are intrinsically related through their effect on the central hydrophobic core of GPCRs. This new unifying general process of activation is consistent with the identification of known constitutively active mutants and an in-depth conservational analysis of significant residues implicated in the process.
Collapse
Affiliation(s)
- Benjamin G Tehan
- Heptares Therapeutics BioPark, Broadwater Road, Welwyn Garden City AL7 3AX United Kingdom.
| | - Andrea Bortolato
- Heptares Therapeutics BioPark, Broadwater Road, Welwyn Garden City AL7 3AX United Kingdom
| | - Frank E Blaney
- Heptares Therapeutics BioPark, Broadwater Road, Welwyn Garden City AL7 3AX United Kingdom
| | - Malcolm P Weir
- Heptares Therapeutics BioPark, Broadwater Road, Welwyn Garden City AL7 3AX United Kingdom
| | - Jonathan S Mason
- Heptares Therapeutics BioPark, Broadwater Road, Welwyn Garden City AL7 3AX United Kingdom
| |
Collapse
|
18
|
Abstract
The understanding of the role of the sphingosine 1-phosphate signaling system in immunology and host defense has deepened exponentially over the past 12 years since the discovery that lymphocyte egress was reversibly modulated by sphingosine 1-phosphate receptors, and with the development of fingolimod, a prodrug of a nonselective S1P receptor agonist, for therapeutic use in the treatment of relapsing, remitting multiple sclerosis. Innovative genetic and chemical approaches, together with structural biology, now provide a more detailed molecular understanding of a regulated lysophospholipid ligand with a variety of autocrine, paracrine, and systemic effects in physiology and pathology, based upon selective interactions with a high affinity and selective evolutionary cluster of G-protein-coupled receptors.
Collapse
|
19
|
Mitigation of radiation injury by selective stimulation of the LPA(2) receptor. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:117-25. [PMID: 23127512 DOI: 10.1016/j.bbalip.2012.08.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/28/2012] [Accepted: 08/31/2012] [Indexed: 11/21/2022]
Abstract
Due to its antiapoptotic action, derivatives of the lipid mediator lysophosphatidic acid (LPA) provide potential therapeutic utility in diseases associated with programmed cell death. Apoptosis is one of the major pathophysiological processes elicited by radiation injury to the organism. Consequently, therapeutic explorations applying compounds that mimic the antiapoptotic action of LPA have begun. Here we present a brief account of our decade-long drug discovery effort aimed at developing LPA mimics with a special focus on specific agonists of the LPA(2) receptor subtype, which was found to be highly effective in protecting cells from apoptosis. We describe new evidence that 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), a prototypic nonlipid agonist specific to the LPA(2) receptor subtype, rescues apoptotically condemned cells in vitro and in vivo from injury caused by high-dose γ-irradiation. GRI977143 shows the features of a radiomitigator because it is effective in rescuing the lives of mice from deadly levels of radiation when administered 24h after radiation exposure. Our findings suggest that by specifically activating LPA(2) receptors GRI977143 activates the ERK1/2 prosurvival pathway, effectively reduces Bax translocation to the mitochondrion, attenuates the activation of initiator and effector caspases, reduces DNA fragmentation, and inhibits PARP-1 cleavage associated with γ-irradiation-induced apoptosis. GRI977143 also inhibits bystander apoptosis elicited by soluble proapoptotic mediators produced by irradiated cells. Thus, GRI977143 can serve as a prototype scaffold for lead optimization paving the way to more potent analogs amenable for therapeutic exploration. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
20
|
O'Sullivan C, Dev KK. The structure and function of the S1P1 receptor. Trends Pharmacol Sci 2013; 34:401-12. [PMID: 23763867 DOI: 10.1016/j.tips.2013.05.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 04/27/2013] [Accepted: 05/07/2013] [Indexed: 12/18/2022]
Abstract
Sphingosine 1-phosphate (S1P) receptors (S1PRs) belong to the class A family of G protein-coupled receptors (GPCRs). S1PRs are widely expressed on many cell types, including those of the immune, cardiovascular, and central nervous systems. The S1PR family is rapidly gaining attention as an important mediator of many cellular processes, including cell differentiation, migration, survival, angiogenesis, calcium homeostasis, inflammation and immunity. Importantly, S1PRs are known drug targets for multiple sclerosis (MS), for which the newly developed oral therapy fingolimod, an S1PR modulator, has recently been approved for clinical use. Much progress has also recently been made in the field of structural biology and in the modeling of heterotrimeric GPCRs allowing the crystal structure of the S1PR1 subtype to be elucidated and key interactions defined. Here, we outline the structure and function of S1PR1, highlighting the key residues involved in receptor activation, signaling, transmembrane interactions, ligand binding, post-translational modification, and protein-protein interactions.
Collapse
Affiliation(s)
- Catherine O'Sullivan
- Molecular Neuropharmacology, Department of Physiology, School of Medicine, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
21
|
Rosen H, Stevens RC, Hanson M, Roberts E, Oldstone MBA. Sphingosine-1-phosphate and its receptors: structure, signaling, and influence. Annu Rev Biochem 2013; 82:637-62. [PMID: 23527695 DOI: 10.1146/annurev-biochem-062411-130916] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The sphingosine-1-phosphate (S1P) receptor signaling system has biological and medical importance and is the first lipid G protein-coupled receptor (GPCR) structure to be solved to 2.8-Å resolution. S1P binds to five high-affinity GPCRs generating multiple downstream signals that play essential roles in vascular development and endothelial integrity, control of cardiac rhythm, and routine oral treatment of multiple sclerosis. Genetics, chemistry, and now structural biology have advanced this integrated biochemical system. The S1P receptors have a novel N-terminal fold that occludes access to the binding pocket from the extracellular environment as well as orthosteric and bitopic ligands with very different physicochemical properties. S1P receptors and metabolizing enzymes have been deleted, inducibly deleted, and knocked in as tagged or altered receptors in mice. An array of genetic models allows analysis of integrated receptor function in vivo. We can now directly understand causal relationships among protein expression, signal, and control points in physiology and pathology.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Chemical Physiology and Immunology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | |
Collapse
|
22
|
Dissecting influenza virus pathogenesis uncovers a novel chemical approach to combat the infection. Virology 2013; 435:92-101. [PMID: 23217619 DOI: 10.1016/j.virol.2012.09.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 09/27/2012] [Accepted: 09/28/2012] [Indexed: 01/09/2023]
Abstract
The cytokine storm is an aggressive immune response characterized by the recruitment of inflammatory leukocytes and exaggerated levels of cytokines and chemokines at the site of infection. Here we review evidence that cytokine storm directly contributes to the morbidity and mortality resulting from influenza virus infection and that sphingosine-1-phosphate (S1P) receptor agonists can abort cytokine storms providing significant protection against pathogenic human influenza viral infections. In experiments using murine models and the human pathogenic 2009 influenza viruses, S1P1 receptor agonist alone reduced deaths from influenza virus by over 80% as compared to lesser protection (50%) offered by the antiviral neuraminidase inhibitor oseltamivir. Optimal protection of 96% was achieved by combined therapy with the S1P1 receptor agonist and oseltamivir. The functional mechanism of S1P receptor agonist(s) action and the predominant role played by pulmonary endothelial cells as amplifiers of cytokine storm during influenza infection are described.
Collapse
|
23
|
Jo E, Bhhatarai B, Repetto E, Guerrero M, Riley S, Brown SJ, Kohno Y, Roberts E, Schürer SC, Rosen H. Novel selective allosteric and bitopic ligands for the S1P(3) receptor. ACS Chem Biol 2012; 7:1975-83. [PMID: 22971058 DOI: 10.1021/cb300392z] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a lysophospholipid signaling molecule that regulates important biological functions, including lymphocyte trafficking and vascular development, by activating G protein-coupled receptors for S1P, namely, S1P(1) through S1P(5). Here, we map the S1P(3) binding pocket with a novel allosteric agonist (CYM-5541), an orthosteric agonist (S1P), and a novel bitopic antagonist (SPM-242). With a combination of site-directed mutagenesis, ligand competition assay, and molecular modeling, we concluded that S1P and CYM-5541 occupy different chemical spaces in the ligand binding pocket of S1P(3). CYM-5541 allowed us to identify an allosteric site where Phe263 is a key gate-keeper residue for its affinity and efficacy. This ligand lacks a polar moiety, and the novel allosteric hydrophobic pocket permits S1P(3) selectivity of CYM-5541 within the highly similar S1P receptor family. However, a novel S1P(3)-selective antagonist, SPM-242, in the S1P(3) pocket occupies the ligand binding spaces of both S1P and CYM-5541, showing its bitopic mode of binding. Therefore, our coordinated approach with biochemical data and molecular modeling, based on our recently published S1P(1) crystal structure data in a highly conserved set of related receptors with a shared ligand, provides a strong basis for the successful optimization of orthosteric, allosteric, and bitopic modulators of S1P(3).
Collapse
Affiliation(s)
- Euijung Jo
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California 92037, United States
| | - Barun Bhhatarai
- Center for Computational Science,
Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Emanuela Repetto
- Control of Gene Expression Laboratory, Batiment Universitaire Archimed, Nice, France
| | - Miguel Guerrero
- Department of Chemistry, The Scripps Research Institute, La Jolla, California
92037, United States
| | - Sean Riley
- The Scripps Research Institute Molecular Screening Center, 10550 North Torrey
Pines Road, La Jolla, California 92037, United States
| | - Steven J. Brown
- The Scripps Research Institute Molecular Screening Center, 10550 North Torrey
Pines Road, La Jolla, California 92037, United States
| | | | - Edward Roberts
- Department of Chemistry, The Scripps Research Institute, La Jolla, California
92037, United States
| | - Stephan C. Schürer
- Center for Computational Science,
Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
- Department of Molecular and Cellular
Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California 92037, United States
- The Scripps Research Institute Molecular Screening Center, 10550 North Torrey
Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
24
|
Kiss GN, Fells JI, Gupte R, Lee SC, Liu J, Nusser N, Lim KG, Ray RM, Lin FT, Parrill AL, Sümegi B, Miller DD, Tigyi G. Virtual screening for LPA2-specific agonists identifies a nonlipid compound with antiapoptotic actions. Mol Pharmacol 2012; 82:1162-73. [PMID: 22968304 PMCID: PMC3502618 DOI: 10.1124/mol.112.079699] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 09/10/2012] [Indexed: 01/11/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a highly potent endogenous lipid mediator that protects and rescues cells from programmed cell death. Earlier work identified the LPA₂ G protein-coupled receptor subtype as an important molecular target of LPA mediating antiapoptotic signaling. Here we describe the results of a virtual screen using single-reference similarity searching that yielded compounds 2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (NSC12404), 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), 4,5-dichloro-2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (H2L5547924), and 2-((9,10-dioxo-9,10-dihydroanthracen-2-yl)carbamoyl) benzoic acid (H2L5828102), novel nonlipid and drug-like compounds that are specific for the LPA₂ receptor subtype. We characterized the antiapoptotic action of one of these compounds, GRI977143, which was effective in reducing activation of caspases 3, 7, 8, and 9 and inhibited poly(ADP-ribose)polymerase 1 cleavage and DNA fragmentation in different extrinsic and intrinsic models of apoptosis in vitro. Furthermore, GRI977143 promoted carcinoma cell invasion of human umbilical vein endothelial cell monolayers and fibroblast proliferation. The antiapoptotic cellular signaling responses were present selectively in mouse embryonic fibroblast cells derived from LPA(1&2) double-knockout mice reconstituted with the LPA₂ receptor and were absent in vector-transduced control cells. GRI977143 was an effective stimulator of extracellular signal-regulated kinase 1/2 activation and promoted the assembly of a macromolecular signaling complex consisting of LPA₂, Na⁺ - H⁺ exchange regulatory factor 2, and thyroid receptor interacting protein 6, which has been shown previously to be a required step in LPA-induced antiapoptotic signaling. The present findings indicate that nonlipid LPA₂-specific agonists represent an excellent starting point for development of lead compounds with potential therapeutic utility for preventing the programmed cell death involved in many types of degenerative and inflammatory diseases.
Collapse
Affiliation(s)
- Gyöngyi N Kiss
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Integrating the puzzle pieces: the current atomistic picture of phospholipid-G protein coupled receptor interactions. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:2-12. [PMID: 22982815 DOI: 10.1016/j.bbalip.2012.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/31/2012] [Accepted: 09/03/2012] [Indexed: 01/09/2023]
Abstract
A compelling question of how phospholipids interact with their target receptors has been of interest since the first receptor-mediated effects were reported. The recent report of a crystal structure for the S1P(1) receptor in complex with an antagonist phospholipid provides interesting perspective on the insights that had previously been gained through structure-activity studies of the phospholipids, as well as modeling and mutagenesis studies of the receptors. This review integrates these varied lines of investigation in the context of their various contributions to our current understanding of phospholipid-receptor interactions. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
26
|
Hanson MA, Roth CB, Jo E, Griffith MT, Scott FL, Reinhart G, Desale H, Clemons B, Cahalan SM, Schuerer SC, Sanna MG, Han GW, Kuhn P, Rosen H, Stevens RC. Crystal structure of a lipid G protein-coupled receptor. Science 2012; 335:851-5. [PMID: 22344443 PMCID: PMC3338336 DOI: 10.1126/science.1215904] [Citation(s) in RCA: 555] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The lyso-phospholipid sphingosine 1-phosphate modulates lymphocyte trafficking, endothelial development and integrity, heart rate, and vascular tone and maturation by activating G protein-coupled sphingosine 1-phosphate receptors. Here, we present the crystal structure of the sphingosine 1-phosphate receptor 1 fused to T4-lysozyme (S1P(1)-T4L) in complex with an antagonist sphingolipid mimic. Extracellular access to the binding pocket is occluded by the amino terminus and extracellular loops of the receptor. Access is gained by ligands entering laterally between helices I and VII within the transmembrane region of the receptor. This structure, along with mutagenesis, agonist structure-activity relationship data, and modeling, provides a detailed view of the molecular recognition and requirement for hydrophobic volume that activates S1P(1), resulting in the modulation of immune and stromal cell responses.
Collapse
Affiliation(s)
- Michael A. Hanson
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | | | - Euijung Jo
- Department of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mark T. Griffith
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | - Fiona L. Scott
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | - Greg Reinhart
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | - Hans Desale
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | - Bryan Clemons
- Receptos, 10835 Road to the Cure, Suite #205, San Diego, CA 92121, USA
| | - Stuart M. Cahalan
- Department of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stephan C. Schuerer
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - M. Germana Sanna
- Department of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Gye Won Han
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Peter Kuhn
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hugh Rosen
- Department of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- The Scripps Research Institute Molecular Screening Center, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
27
|
Recent Advances in the Discovery and Development of Sphingosine-1-Phosphate-1 Receptor Agonists. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2012. [DOI: 10.1016/b978-0-12-396492-2.00013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
28
|
Abstract
Comparative modeling is a powerful technique to generate models of proteins from families already represented by members with experimentally characterized three-dimensional structures. The method is particularly important for modeling membrane-bound receptors in the G Protein-Coupled Receptor (GPCR) family, such as many of the lipid receptors (such as the cannabinoid, prostanoid, lysophosphatidic acid, sphingosine 1-phosphate, and eicosanoid receptor family members), as these represent particularly challenging targets for experimental structural characterization methods. Although challenging modeling targets, these receptors have been linked to therapeutic indications that vary from nociception to cancer, and thus are of interest as therapeutic targets. Accurate models of lipid receptors are therefore valuable tools in the drug discovery and optimization phases of therapeutic development. This chapter describes the construction and evaluation of comparative structural models of lipid receptors beginning with the selection of template structures.
Collapse
Affiliation(s)
- Abby L Parrill
- Department of Chemistry, The University of Memphis, Memphis, TN, USA.
| |
Collapse
|
29
|
Pennington LD, Sham KKC, Pickrell AJ, Harrington PE, Frohn MJ, Lanman BA, Reed AB, Croghan MD, Lee MR, Xu H, McElvain M, Xu Y, Zhang X, Fiorino M, Horner M, Morrison HG, Arnett HA, Fotsch C, Wong M, Cee VJ. 4-Methoxy-N-[2-(trifluoromethyl)biphenyl-4-ylcarbamoyl]nicotinamide: A Potent and Selective Agonist of S1P1. ACS Med Chem Lett 2011; 2:752-7. [PMID: 24900263 DOI: 10.1021/ml2001399] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 07/29/2011] [Indexed: 01/06/2023] Open
Abstract
The sphingosine-1-phosphate-1 receptor (S1P1) and its endogenous ligand sphingosine-1-phosphate (S1P) cooperatively regulate lymphocyte trafficking from the lymphatic system. Herein, we disclose 4-methoxy-N-[2-(trifluoromethyl)biphenyl-4-ylcarbamoyl]nicotinamide (8), an uncommon example of a synthetic S1P1 agonist lacking a polar headgroup, which is shown to effect dramatic reduction of circulating lymphocytes (POC = -78%) in rat 24 h after a single oral dose (1 mg/kg). The excellent potency that 8 exhibits toward S1P1 (EC50 = 0.035 μM, 96% efficacy) and the >100-fold selectivity that it displays against receptor subtypes S1P2-5 suggest that it may serve as a valuable tool to understand the clinical relevance of selective S1P1 agonism.
Collapse
Affiliation(s)
- Lewis D. Pennington
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Kelvin K. C. Sham
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Alexander J. Pickrell
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Paul E. Harrington
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Michael J. Frohn
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Brian A. Lanman
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Anthony B. Reed
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Michael D. Croghan
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Matthew R. Lee
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Han Xu
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Michele McElvain
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Yang Xu
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Xuxia Zhang
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Michael Fiorino
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Michelle Horner
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Henry G. Morrison
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Heather A. Arnett
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Christopher Fotsch
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Min Wong
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Victor J. Cee
- Medicinal Chemistry, ‡Molecular Structure, §HTS and Molecular Pharmacology, ∥Inflammation Research, ⊥Pharmacokinetics and Drug Metabolism, #Toxicology, and +Pharmaceutics, Amgen, One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| |
Collapse
|
30
|
Blaho VA, Hla T. Regulation of mammalian physiology, development, and disease by the sphingosine 1-phosphate and lysophosphatidic acid receptors. Chem Rev 2011; 111:6299-320. [PMID: 21939239 PMCID: PMC3216694 DOI: 10.1021/cr200273u] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Victoria A. Blaho
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY 10065
| | - Timothy Hla
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY 10065
| |
Collapse
|
31
|
Agonist-dependent effects of mutations in the sphingosine-1-phosphate type 1 receptor. Eur J Pharmacol 2011; 667:105-12. [DOI: 10.1016/j.ejphar.2011.05.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/05/2011] [Accepted: 05/22/2011] [Indexed: 11/17/2022]
|
32
|
Valentine WJ, Godwin VI, Osborne DA, Liu J, Fujiwara Y, Van Brocklyn J, Bittman R, Parrill AL, Tigyi G. FTY720 (Gilenya) phosphate selectivity of sphingosine 1-phosphate receptor subtype 1 (S1P1) G protein-coupled receptor requires motifs in intracellular loop 1 and transmembrane domain 2. J Biol Chem 2011; 286:30513-30525. [PMID: 21719706 DOI: 10.1074/jbc.m111.263442] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
FTY720 phosphate (FTY720P) is a high potency agonist for all the endothelial differentiation gene family sphingosine 1-phosphate (S1P) receptors except S1P receptor subtype 2 (S1P(2)). To map the distinguishing features of S1P(2) ligand recognition, we applied a computational modeling-guided mutagenesis strategy that was based on the high degree of sequence homology between S1P(1) and S1P(2). S1P(2) point mutants of the ligand-binding pocket were characterized. The head group-interacting residues Arg3.28, Glu3.29, and Lys7.34 were essential for activation. Mutation of residues Ala3.32, Leu3.36, Val5.41, Phe6.44, Trp6.48, Ser7.42, and Ser7.46, predicted to interact with the S1P hydrophobic tail, impaired activation by S1P. Replacing individual or multiple residues in the ligand-binding pocket of S1P(2) with S1P(1) sequence did not impart activation by FTY720P. Chimeric S1P(1)/S1P(2) receptors were generated and characterized for activation by S1P or FTY720P. The S1P(2) chimera with S1P(1) sequence from the N terminus to transmembrane domain 2 (TM2) was activated by FTY720P, and the S1P(2)(IC1-TM2)(S1P1) domain insertion chimera showed S1P(1)-like activation. Twelve residues in this domain, distributed in four motifs a-d, differ between S1P(1) and S1P(2). Insertion of (78)RPMYY in motif b alone or simultaneous swapping of five other residues in motifs c and d from S1P(1) into S1P(2) introduced FTY720P responsiveness. Molecular dynamics calculations indicate that FTY720P binding selectivity is a function of the entropic contribution to the binding free energy rather than enthalpic contributions and that preferred agonists retain substantial flexibility when bound. After exposure to FTY720P, the S1P(2)(IC1-TM2)(S1P1) receptor recycled to the plasma membrane, indicating that additional structural elements are required for the selective degradative trafficking of S1P(1).
Collapse
Affiliation(s)
- William J Valentine
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Virginia I Godwin
- Department of Chemistry, Computational Research on Materials Institute, University of Memphis, Memphis, Tennessee 38152
| | - Daniel A Osborne
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Jianxiong Liu
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Yuko Fujiwara
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of The City University of New York, Flushing, New York 11367-1597
| | - Abby L Parrill
- Department of Chemistry, Computational Research on Materials Institute, University of Memphis, Memphis, Tennessee 38152
| | - Gabor Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163.
| |
Collapse
|
33
|
Pyne S, Pyne NJ. Translational aspects of sphingosine 1-phosphate biology. Trends Mol Med 2011; 17:463-72. [PMID: 21514226 DOI: 10.1016/j.molmed.2011.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 02/28/2011] [Accepted: 03/04/2011] [Indexed: 12/16/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that has both physiological and pathophysiological roles. It regulates cellular processes such as proliferation, migration, survival and differentiation and affects all organ systems. S1P not only activates S1P-specific receptors to initiate cellular signalling pathways but also directly regulates specific intracellular target proteins. The therapeutic opportunities surrounding S1P signalling are numerous and exemplified by the recent approval of FTY720 (a sphingosine analogue, Gilenya™) for the treatment of relapsing multiple sclerosis. A major focus of research is to develop small-molecule antagonists/agonists/inhibitors that are specific to the different S1P receptor subtypes and the enzymes that regulate S1P levels. This review describes fundamental aspects of S1P biology with an emphasis on the translational potential of intervention therapeutics.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | | |
Collapse
|
34
|
Pyrazole derived from (+)-3-carene; a novel potent, selective scaffold for sphingosine-1-phosphate (S1P1) receptor agonists. Bioorg Med Chem Lett 2010; 20:35-7. [DOI: 10.1016/j.bmcl.2009.11.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 11/11/2009] [Accepted: 11/12/2009] [Indexed: 11/22/2022]
|
35
|
Li G, Mosier PD, Fang X, Zhang Y. Toward the three-dimensional structure and lysophosphatidic acid binding characteristics of the LPA(4)/p2y(9)/GPR23 receptor: a homology modeling study. J Mol Graph Model 2009; 28:70-9. [PMID: 19423373 DOI: 10.1016/j.jmgm.2009.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 03/24/2009] [Accepted: 04/11/2009] [Indexed: 11/16/2022]
Abstract
Lysophosphatidic acid (LPA) is a naturally occurring phospholipid that initiates a broad array of biological processes, including those involved in cell proliferation, survival and migration via activation of specific G protein-coupled receptors located on the cell surface. To date, at least five receptor subtypes (LPA(1-5)) have been identified. The LPA(1-3) receptors are members of the endothelial cell differentiation gene (Edg) family. LPA(4), a member of the purinergic receptor family, and the recently identified LPA(5) are structurally distant from the canonical Edg LPA(1-3) receptors. LPA(4) and LPA(5) are linked to G(q), G(12/13) and G(s) but not G(i), while LPA(1-3) all couple to G(i) in addition to G(q) and G(12/13). There is also evidence that LPA(4) and LPA(5) are functionally different from the Edg LPA receptors. Computational modeling has provided useful information on the structure-activity relationship (SAR) of the Edg LPA receptors. In this work, we focus on the initial analysis of the structural and ligand-binding properties of LPA(4), a prototype non-Edg LPA receptor. Three homology models of the LPA(4) receptor were developed based on the X-ray crystal structures of the ground state and photoactivated bovine rhodopsin and the recently determined human beta(2)-adrenergic receptor. Docking studies of LPA in the homology models were then conducted, and plausible LPA binding loci were explored. Based on these analyses, LPA is predicted to bind to LPA(4) in an orientation similar to that reported for LPA(1-3), but through a different network of hydrogen bonds. In LPA(1-3), the ligand polar head group is reported to interact with residues at positions 3.28, 3.29 and 7.36, whereas three non-conserved amino acid residues, S114(3.28), T187(EL2) and Y265(6.51), are predicted to interact with the polar head group in the LPA(4) receptor models.
Collapse
Affiliation(s)
- Guo Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298-0540, USA
| | | | | | | |
Collapse
|
36
|
Straub AC, Klei LR, Stolz DB, Barchowsky A. Arsenic requires sphingosine-1-phosphate type 1 receptors to induce angiogenic genes and endothelial cell remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1949-58. [PMID: 19349368 DOI: 10.2353/ajpath.2009.081016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Arsenic in drinking water is a major public health concern as it increases risk and incidence of cardiovascular disease and cancer. Arsenic exposure affects multiple vascular beds, promoting liver sinusoidal capillarization and portal hypertension, ischemic heart disease, peripheral vascular disease, and tumor angiogenesis. While Rac1-GTPase and NADPH oxidase activities are essential for arsenic-stimulated endothelial cell signaling for angiogenesis or liver sinusoid capillarization, the mechanism for initiating these effects is unknown. We found that arsenic-stimulated cell signaling and angiogenic gene expression in human microvascular endothelial cells were Pertussis toxin sensitive, indicating a G-protein coupled signaling pathway. Incubating human microvascular endothelial cells with the sphingosine-1-phosphate type 1 receptor (S1P(1)) inhibitor VPC23019 or performing small interfering RNA knockdown of S1P(1) blocked arsenic-stimulated HMVEC angiogenic gene expression and tube formation, but did not affect induction of either HMOX1 or IL8. Liver sinusoidal endothelial cells (LSECs) defenestrate and capillarize in response to aging and environmental oxidant stresses. We found that S1P(1) was enriched on LSECs in vivo and in primary cell culture and that VPC23019 inhibited both sphingosine-1-phosphate-stimulated and arsenic-stimulated LSEC oxidant generation and defenestration. These studies identified novel roles for S1P(1) in mediating arsenic stimulation of both angiogenesis and pathogenic LSEC capillarization, as well as demonstrating a role for S1P(1) in mediating environmental responses in the liver vasculature, providing possible mechanistic insight into arsenic-induced vascular pathogenesis and disease.
Collapse
Affiliation(s)
- Adam C Straub
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | | | | | | |
Collapse
|
37
|
Liao JJ, Huang MC, Fast K, Gundling K, Yadav M, Van Brocklyn JR, Wabl MR, Goetzl EJ. Immunosuppressive human anti-lymphocyte autoantibodies specific for the type 1 sphingosine 1-phosphate receptor. FASEB J 2009; 23:1786-96. [PMID: 19158154 DOI: 10.1096/fj.08-124891] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Anti-lymphocyte antibodies (Abs) that suppress T-cell chemotactic and other responses to sphingosine 1-phosphate (S1P), but not to chemokines, were found in a lymphopenic patient with recurrent infections. Lymphocyte type 1 S1P receptor (S1P(1)) that transduces S1P chemotactic stimulation was recognized by patient Abs in Western blots of T cells, S1P(1) transfectants, and S1P(1)-hemagglutinin purified by monoclonal anti-hemagglutinin Ab absorption. The amino terminus of S1P(1), but not any extracellular loop, prevented anti-S1P(1) Ab suppression of S1P(1) signaling and T-cell chemotaxis to S1P. Human purified anti-S1P(1) Abs decreased mouse blood lymphocyte levels by a mean of 72%, suppressed mouse T-cell chemotaxis to S1P in vivo, and significantly reduced the severity of dextran sodium sulfate-induced colitis in mice. Human Abs to the amino terminus of S1P(1) suppress T-cell trafficking sufficiently to impair host defense and provide therapeutic immunosuppression.
Collapse
Affiliation(s)
- Jia-Jun Liao
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Chapter 12 The Use of Receptor Homology Modeling to Facilitate the Design of Selective Chemokine Receptor Antagonists. Methods Enzymol 2009; 461:249-79. [DOI: 10.1016/s0076-6879(09)05412-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
Gonzalez-Cabrera PJ, Jo E, Sanna MG, Brown S, Leaf N, Marsolais D, Schaeffer MT, Chapman J, Cameron M, Guerrero M, Roberts E, Rosen H. Full pharmacological efficacy of a novel S1P1 agonist that does not require S1P-like headgroup interactions. Mol Pharmacol 2008; 74:1308-18. [PMID: 18708635 PMCID: PMC2575047 DOI: 10.1124/mol.108.049783] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Strong evidence exists for interactions of zwitterionic phosphate and amine groups in sphingosine-1 phosphate (S1P) to conserved Arg and Glu residues present at the extracellular face of the third transmembrane domain of S1P receptors. The contribution of Arg(120) and Glu(121) for high-affinity ligand-receptor interactions is essential, because single-point R(120)A or E(121)A S1P(1) mutants neither bind S1P nor transduce S1P function. Because S1P receptors are therapeutically interesting, identifying potent selective agonists with different binding modes and in vivo efficacy is of pharmacological importance. Here we describe a modestly water-soluble highly selective S1P(1) agonist [2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino) ethanol (CYM-5442)] that does not require Arg(120) or Glu(121) residues for activating S1P(1)-dependent p42/p44 mitogen-activated protein kinase phosphorylation, which defines a new hydrophobic pocket in S1P(1). CYM-5442 is a full agonist in vitro for S1P(1) internalization, phosphorylation, and ubiquitination. It is noteworthy that CYM-5442 was a full agonist for induction and maintenance of S1P(1)-dependent blood lymphopenia, decreasing B lymphocytes by 65% and T lymphocytes by 85% of vehicle. Induction of CYM-5442 lymphopenia was dose- and time-dependent, requiring serum concentrations in the 50 nM range. In vitro measures of S1P(1) activation by CYM-5442 were noncompetitively inhibited by a specific S1P(1) antagonist [(R)-3-amino-(3-hexylphenylamino)-4-oxobutylphosphonic acid (W146)], competitive for S1P, 2-amino-2-(4-octylphenethyl)propane-1,3-diol (FTY720-P), and 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2, 4-oxadiazole (SEW2871). In addition, lymphopenia induced by CYM-5442 was reversed by W146 administration or upon pharmacokinetic agonist clearance. Pharmacokinetics in mice also indicated that CYM-5442 partitions significantly in central nervous tissue. These data show that CYM-5442 activates S1P(1)-dependent pathways in vitro and to levels of full efficacy in vivo through a hydrophobic pocket separate from the orthosteric site of S1P binding that is headgroup-dependent.
Collapse
Affiliation(s)
| | | | | | | | - Nora Leaf
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - David Marsolais
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - Marie-Therese Schaeffer
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - Jacqueline Chapman
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - Michael Cameron
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - Miguel Guerrero
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | - Edward Roberts
- Departments of Chemical Physiology & Immunology (PJG-C, EJ, MGS, NL, DM, HR), Chemistry (MG, ER) and The Scripps Research Institute Molecular Screening Center (M-TS,JC,SB, HR), 10550 North Torrey Pines Rd, La Jolla, CA 92037, Translational Research Institute, Scripps Florida, 5353 Parkside Drive, Jupiter, FL 33458 (MC)
| | | |
Collapse
|
40
|
Parrill AL. Lysophospholipid interactions with protein targets. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1781:540-6. [PMID: 18501204 PMCID: PMC2564851 DOI: 10.1016/j.bbalip.2008.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 04/22/2008] [Accepted: 04/23/2008] [Indexed: 12/21/2022]
Abstract
Bioactive lysophospholipids include lysophosphatidic acid (LPA), sphingosine 1-phosphate (S1P), cyclic-phosphatidic acid (CPA) and alkyl glycerolphosphate (AGP). These lipid mediators stimulate a variety of responses that include cell survival, proliferation, migration, invasion, wound healing, and angiogenesis. Responses to lysophospholipids depend upon interactions with biomolecular targets in the G protein-coupled receptor (GPCR) and nuclear receptor families, as well as enzymes. Our current understanding of lysophospholipid interactions with these targets is based on a combination of lysophospholipid analog structure activity relationship studies as well as more direct structural characterization techniques such as X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and experimentally-validated molecular modeling. The direct structural characterization studies are the focus of this review, and provide the insight necessary to stimulate structure-based therapeutic lead discovery efforts in the future.
Collapse
Affiliation(s)
- Abby L Parrill
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, USA.
| |
Collapse
|
41
|
Schürer SC, Brown SJ, Cabrera PG, Schaeffer MT, Chapman J, Jo E, Chase P, Spicer T, Hodder P, Rosen H. Ligand-binding pocket shape differences between sphingosine 1-phosphate (S1P) receptors S1P1 and S1P3 determine efficiency of chemical probe identification by ultrahigh-throughput screening. ACS Chem Biol 2008; 3:486-98. [PMID: 18590333 PMCID: PMC2597349 DOI: 10.1021/cb800051m] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We have studied the sphingosine 1-phosphate (S1P) receptor system to better understand why certain molecular targets within a closely related family are much more tractable when identifying compelling chemical leads. Five medically important G-protein-coupled receptors for S1P regulate heart rate, coronary artery caliber, endothelial barrier integrity, and lymphocyte trafficking. Selective S1P receptor agonist probes would be of great utility to study receptor subtype-specific function. Through systematic screening of the same libraries, we identified novel selective agonist chemotypes for each of the S1P1 and S1P3 receptors. Ultrahigh-throughput screening (uHTS) for S1P1 was more effective than that for S1P3, with many selective, low nanomolar hits of proven mechanism emerging. Receptor structure modeling and ligand docking reveal differences between the receptor binding pockets, which are the basis for subtype selectivity. Novel selective agonists interact primarily in the hydrophobic pocket of the receptor in the absence of headgroup interactions. Chemistry-space and shape-based analysis of the screening libraries in combination with the binding models explain the observed differential hit rates and enhanced efficiency for lead discovery for S1P1 versus S1P3 in this closely related receptor family.
Collapse
Affiliation(s)
- Stephan C. Schürer
- Department of Scientific Computing, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Steven J. Brown
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Pedro Gonzales Cabrera
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Marie-Therese Schaeffer
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Jacqueline Chapman
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Euijung Jo
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Peter Chase
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Tim Spicer
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Peter Hodder
- The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California and Jupiter, Florida
| |
Collapse
|
42
|
Truc-Chi T, Fells JI, Osborne DA, North EJ, Naor MM, Parrill AL. Molecular recognition in the sphingosine 1-phosphate receptor family. J Mol Graph Model 2008; 26:1189-201. [PMID: 18165127 PMCID: PMC2413001 DOI: 10.1016/j.jmgm.2007.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 11/01/2007] [Accepted: 11/03/2007] [Indexed: 10/22/2022]
Abstract
Computational modeling and its application in ligand screening and ligand receptor interaction studies play important roles in structure-based drug design. A series of sphingosine 1-phosphate (S1P) receptor ligands with varying potencies and receptor selectivities were docked into homology models of the S1P(1-5) receptors. These studies provided molecular insights into pharmacological trends both across the receptor family as well as at single receptors. This study identifies ligand recognition features that generalize across the S1P receptor family, features unique to the S1P(4) and S1P(5) receptors, and suggests significant structural differences of the S1P(2) receptor. Docking results reveal a previously unknown sulfur-aromatic interaction between the S1P(4) C5.44 sulfur atom and the phenyl ring of benzimidazole as well as pi-pi interaction between F3.33 of S1P(1,4,5) and aromatic ligands. The findings not only confirm the importance of a cation-pi interaction between W4.64 and the ammonium of S1P at S1P(4) but also predict the same interaction at S1P(5). S1P receptor models are validated for pharmacophore development including database mining and new ligand discovery and serve as tools for ligand optimization to improve potency and selectivity.
Collapse
Affiliation(s)
- T. Truc-Chi
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - James I. Fells
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - Daniel A. Osborne
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - E. Jeffrey North
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - Mor M. Naor
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - Abby L. Parrill
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| |
Collapse
|
43
|
Fells JI, Tsukahara R, Fujiwara Y, Liu J, Perygin DH, Osborne DA, Tigyi G, Parrill AL. Identification of non-lipid LPA3 antagonists by virtual screening. Bioorg Med Chem 2008; 16:6207-17. [PMID: 18467108 PMCID: PMC2483252 DOI: 10.1016/j.bmc.2008.04.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 04/15/2008] [Accepted: 04/16/2008] [Indexed: 10/22/2022]
Abstract
In the present study, we utilized virtual screening to identify LPA(3) antagonists. We have developed a three-point structure-based pharmacophore model based on known LPA(3) antagonists. This model was used to mine the NCI database. Docking, pharmacophore development, and database mining produced new, non-lipid leads. Experimental testing of seven computationally selected pharmacophore hits produced one potentiator and three antagonists, one of which displays both LPA(3) selectivity and nanomolar potency. Similarity searching in the ChemBridge database using the most promising lead as the search target produced four additional LPA(3) antagonists and a potent dual LPA(1&2) antagonist.
Collapse
Affiliation(s)
- James I. Fells
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152
| | - Ryoko Tsukahara
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Yuko Fujiwara
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Jianxiong Liu
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Donna H. Perygin
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152
| | - Daniel A. Osborne
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Abby L. Parrill
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152
| |
Collapse
|
44
|
Valentine WJ, Fells JI, Perygin DH, Mujahid S, Yokoyama K, Fujiwara Y, Tsukahara R, Van Brocklyn JR, Parrill AL, Tigyi G. Subtype-specific residues involved in ligand activation of the endothelial differentiation gene family lysophosphatidic acid receptors. J Biol Chem 2008; 283:12175-87. [PMID: 18316373 DOI: 10.1074/jbc.m708847200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a ligand for three endothelial differentiation gene family G protein-coupled receptors, LPA(1-3). We performed computational modeling-guided mutagenesis of conserved residues in transmembrane domains 3, 4, 5, and 7 of LPA(1-3) predicted to interact with the glycerophosphate motif of LPA C18:1. The mutants were expressed in RH7777 cells, and the efficacy (E(max)) and potency (EC(50)) of LPA-elicited Ca(2+) transients were measured. Mutation to alanine of R3.28 universally decreased both the efficacy and potency in LPA(1-3) and eliminated strong ionic interactions in the modeled LPA complexes. The alanine mutation at Q3.29 decreased modeled interactions and activation in LPA(1) and LPA(2) more than in LPA(3). The mutation W4.64A had no effect on activation and modeled LPA interaction of LPA(1) and LPA(2) but reduced the activation and modeled interactions of LPA(3). The R5.38A mutant of LPA(2) and R5.38N mutant of LPA(3) showed diminished activation by LPA; however, in LPA(1) the D5.38A mutation did not, and mutation to arginine enhanced receptor activation. In LPA(2), K7.36A decreased the potency of LPA; in LPA(1) this same mutation increased the E(max). In LPA(3), R7.36A had almost no effect on receptor activation; however, the mutation K7.35A increased the EC(50) in response to LPA 10-fold. In LPA(1-3), the mutation Q3.29E caused a modest increase in EC(50) in response to LPA but caused the LPA receptors to become more responsive to sphingosine 1-phosphate (S1P). Surprisingly micromolar concentrations of S1P activated the wild type LPA(2) and LPA(3) receptors, indicating that S1P may function as a weak agonist of endothelial differentiation gene family LPA receptors.
Collapse
Affiliation(s)
- William J Valentine
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Naor MM, Walker MD, Van Brocklyn JR, Tigyi G, Parrill AL. Sphingosine 1-phosphate pKa and binding constants: intramolecular and intermolecular influences. J Mol Graph Model 2007; 26:519-28. [PMID: 17467317 PMCID: PMC2040500 DOI: 10.1016/j.jmgm.2007.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/10/2007] [Accepted: 03/12/2007] [Indexed: 11/25/2022]
Abstract
The dissociation constant for an ionizable ligand binding to a receptor is dependent on its charge and therefore on its environmentally-influenced pKa value. The pKa values of sphingosine 1-phosphate (S1P) were studied computationally in the context of the wild type S1P1 receptor and the following mutants: E3.29Q, E3.29A, and K5.38A. Calculated pKa values indicate that S1P binds to S1P1 and its site mutants with a total charge of -1, including a +1 charge on the ammonium group and a -2 charge on the phosphate group. The dissociation constant of S1P binding to these receptors was studied as well. The models of wild type and mutant proteins originated from an active receptor model that was developed previously. We used ab initio RHF/6-31+G(d) to optimize our models in aqueous solution, where the solvation energy derivatives are represented by conductor-like polarizable continuum model (C-PCM) and integral equation formalism polarizable continuum model (IEF-PCM). Calculation of the dissociation constant for each mutant was determined by reference to the experimental dissociation constant of the wild type receptor. The computed dissociation constants of the E3.29Q and E3.29A mutants are three to five orders of magnitude higher than those for the wild type receptor and K5.38A mutant, indicating vital contacts between the S1P phosphate group and the carboxylate group of E3.29. Computational dissociation constants for K5.38A, E3.29A, and E3.29Q mutants were compared with experimentally determined binding and activation data. No measurable binding of S1P to the E3.29A and E3.29Q mutants was observed, supporting the critical contacts observed computationally. These results validate the quantitative accuracy of the model.
Collapse
Affiliation(s)
- Mor M. Naor
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - Michelle D. Walker
- Department of Physiology and University of Tennessee Cancer Institute, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - James R. Van Brocklyn
- Division of Neuropathology, Department of Pathology, The Ohio State University, Columbus, Ohio 43210
| | - Gabor Tigyi
- Department of Physiology and University of Tennessee Cancer Institute, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Abby L. Parrill
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| |
Collapse
|
46
|
Molderings GJ, Bönisch H, Brüss M, Wolf C, von Kügelgen I, Göthert M. S1P-receptors in PC12 and transfected HEK293 cells: molecular targets of hypotensive imidazoline I(1) receptor ligands. Neurochem Int 2007; 51:476-85. [PMID: 17559976 DOI: 10.1016/j.neuint.2007.04.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/23/2007] [Accepted: 04/27/2007] [Indexed: 02/07/2023]
Abstract
The present study aimed at elucidating the molecular identity of the proposed "I(1)-imidazoline receptors", i.e. non-adrenoceptor recognition sites via which the centrally acting imidazolines clonidine and moxonidine mediate a major part of their effects. In radioligand binding experiments with [(3)H]clonidine and [(3)H]lysophosphatidic acid on intact, alpha(2)-adrenoceptor-deficient PC12 cells, moxonidine, clonidine, lysophosphatidic acid and sphingosine-1-phosphate (S1P) competed for the specific binding sites of both radioligands with similar affinities. RNA interference with the rat S1P(1)-, S1P(2)- or S1P(3)-receptor abolished specific [(3)H]lysophosphatidic acid binding. [(3)H]Clonidine binding was markedly decreased by siRNA targeting S1P(1)- and S1P(3)-receptors but not by siRNA against S1P(2)-receptors. Finally, in HEK293 cells transiently expressing human S1P(3)-receptors, sphingosine-1-phosphate, clonidine and moxonidine induced increases in intracellular calcium concentration, moxonidine being more potent than clonidine; this is in agreement with the known properties of the "I(1)-imidazoline receptors". The present results indicate that the "I(1)-imidazoline receptors" mediating effects of clonidine and moxonidine in PC12 and the transfected HEK293 cells belong to the S1P-receptor family; in particular, the data obtained in PC12 cells suggest that the I(1) imidazoline receptors represent a mixture of S1P(1)- and S1P(3)-receptors and/or hetero-dimers of both.
Collapse
Affiliation(s)
- Gerhard J Molderings
- University of Bonn, Institute of Pharmacology and Toxicology, Reuterstr. 2b, D-53113 Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Chapter 16 Sphingosine 1-Phosphate Type 1 Receptor Modulators: Recent Advances and Therapeutic Potential. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2007. [DOI: 10.1016/s0065-7743(07)42016-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|